1
|
Abstract
Bone marrow, in addition to hematopoietic precursors, contains cells that are considered stem cells of nonhematopoietic tissues. These cells are referred to as marrow stromal cells or mesenchymal stem cells. Marrow stromal cells, because of their ability to survive, integrate, and migrate within the central nervous system, can be used as an alternative source of cells for neural transplantation and repair. They can be expanded rapidly in culture and can be induced to express markers of neural cells. Moreover, implanted into the developing brain, these cells can integrate without disrupting the host brain architecture and can assume the fate of neural cells. They can be genetically transduced and can elaborate transgene products. Because large numbers of stromal cells can be obtained from small aspirates of bone marrow, these cells are potentially useful for treating a variety of neurological diseases.
Collapse
Affiliation(s)
- S. Ausim Azizi
- Department of Neurology and Center for Gene Therapy, MCP-Hahnemann University, Philadelphia, Pennsylvania,
| |
Collapse
|
2
|
Abstract
It is possible, by a variety of means, to isolate, propagate, and characterize engraftable clones of cells from the mammalian CNS that fulfill the operational definition of a "stem cell": self-maintaining, self-renewing, and extremely multipotent in vitro and in vivo. Even as debates flourish over how neural stem cells might best be defined, identified, represented, and manipulated, clonal cells with "stem-like" features have begun to provide valuable models for studying commitment, differentiation, and plasticity in the CNS. Furthermore, by learning in this way the basic biology of neural stem cells, and by then exploiting those inherent properties for therapeutic ends, novel and multifaceted strategies seem poised to emerge for redressing a variety of heretofore untreatable CNS dysfunctions. Stem-like cells have begun to show promise for neural cell re placement and molecular support therapy in various animal models of degenerative, developmental, and acquired CNS insult. NEUROSCIENTIST 4:408-425, 1998
Collapse
Affiliation(s)
- Evan Y. Snyder
- Departments of Neurology (Division of Neuroscience) and Pediatrics (Division of Newborn Medicine) Harvard Medical School Children's Hospital Boston, Massachusetts
| |
Collapse
|
3
|
Jalali H, Parivar K, Soleimani M, Nabiuni M, Aghaee-Bakhtiari H. Ex-Vivo Gene Therapy Using Lentiviral Mediated Gene Transfer Into Umbilical Cord Blood Derived Stem Cells. ACTA ACUST UNITED AC 2016. [DOI: 10.17795/zjrms-5991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
4
|
Shakhbazau A, Mohanty C, Kumar R, Midha R. Sensory recovery after cell therapy in peripheral nerve repair: effects of naïve and skin precursor-derived Schwann cells. J Neurosurg 2014; 121:423-31. [DOI: 10.3171/2014.5.jns132132] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Object
Cell therapy is a promising candidate among biological or technological innovations sought to augment microsurgical techniques in peripheral nerve repair. This report describes long-term functional regenerative effects of cell therapy in the rat injury model with a focus on sensory recovery.
Methods
Schwann cells were derived from isogenic nerve or skin precursor cells and injected into the transected and immediately repaired sciatic nerve distal to the injury site. Sensory recovery was assessed at weeks 4, 7, and 10. Axonal regeneration was assessed at Week 11.
Results
By Week 10, thermal sensitivity in cell therapy groups returned to a level indistinguishable from the baseline (p > 0.05). Immunohistochemistry at 11 weeks after injury showed improved regeneration of NF+ and IB4+ axons.
Conclusions:
The results of this study show that cell therapy significantly improves thermal sensation and the number of regenerated sensory neurons at 11 weeks after injury. These findings contribute to the view of skin-derived stem cells as a reliable source of Schwann cells with therapeutic potential for functional recovery in damaged peripheral nerve.
Collapse
Affiliation(s)
- Antos Shakhbazau
- 1Department of Clinical Neuroscience, Faculty of Medicine,
- 2Hotchkiss Brain Institute, and
| | | | - Ranjan Kumar
- 1Department of Clinical Neuroscience, Faculty of Medicine,
- 2Hotchkiss Brain Institute, and
- 3Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rajiv Midha
- 1Department of Clinical Neuroscience, Faculty of Medicine,
- 2Hotchkiss Brain Institute, and
| |
Collapse
|
5
|
Jalali H, Parivar K, Nabiuni M, Soleimani M. Unrestricted somatic stem cells as vehicle for nerve growth factor gene transfer. Neurol Res 2013; 35:553-560. [PMID: 23561736 DOI: 10.1179/1743132813y.0000000176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Nerve growth factor (NGF), a member of the neurotrophic factor family, plays a critical role in the maintenance and regeneration of different types of neurons. To overcome drastic challenges in the peripheral delivery of NGF, transplantation of NGF secreting stem cells to the target site of an injury may be an effective procedure. Unrestricted somatic stem cells (USSCs), a subtype of umbilical cord blood (UCB) stem cells, have shown promise for gene therapy purposes, and proper results have been observed from transplantation experiments in neurodegenerative disorders. Based on the considerable potential of USSCs for gene delivery applications, the goal of the current study was to establish a betaNGF gene containing USSCs, which is able to secrete functional recombinant betaNGF protein. METHODS Unrestricted somatic stem cells were isolated from UCB and were cultured in a DMEM medium. The betaNGF gene was cloned in the EFalpha-promoting lentiviral vector, and virus production was performed as a third generation lentivirus packaging system. Enzyme-linked immunosorbent assay, real-time PCR, co-culture, MTT, and immunocytochemistry assays were performed to evaluate the genetically engineered USSCs. RESULTS Overexpression of betaNGF gene in human USSCs created a USSC line that is able to secrete high amounts of functional betaNGF protein. betaNGF-secreting USSCs showed a high rate of viability along with acceptable immunological and morphological properties for transplantation into the nervous system. DISCUSSION Long-term expression of functional betaNGF, high viability of betaNGF producing USSCs, and expression of primary neuronal markers suggest that USSCs may be useful to deliver betaNGF into targeted sites of the nervous system in neurodegenerative disorders.
Collapse
Affiliation(s)
- Hanieh Jalali
- Department of Developmental Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran.
| | | | | | | |
Collapse
|
6
|
Early regenerative effects of NGF-transduced Schwann cells in peripheral nerve repair. Mol Cell Neurosci 2012; 50:103-12. [PMID: 22735691 DOI: 10.1016/j.mcn.2012.04.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 03/27/2012] [Accepted: 04/06/2012] [Indexed: 01/04/2023] Open
Abstract
Peripheral nerve injury leads to a rapid and robust increase in the synthesis of neurotrophins which guide and support regenerating axons. To further optimize neurotrophin supply at the earliest stages of regeneration, we over-expressed NGF in Schwann cells (SCs) by transducing these cells with a lentiviral vector encoding NGF (NGF-SCs). Transplantation of NGF-SCs in a rat sciatic nerve transection/repair model led to significant increase of NGF levels 2weeks after injury and correspondingly to substantial improvement in axonal regeneration. Numbers of NF200, ChAT and CGRP-positive axon profiles, as well as the gastrocnemius muscle weights, were significantly higher in the NGF-Schwann cell group compared to the animals that received control SCs transduced with a lentiviral vector encoding GFP (GFP-SCs). Comparison with other models of NGF application signifies the important role of this neurotrophin during the early stages of regeneration, and supports the importance of developing combined gene and cell therapy for peripheral nerve repair.
Collapse
|
7
|
Foamy virus: an available vector for gene transfer in neural cells and other nondividing cells. J Neurovirol 2010; 16:419-26. [DOI: 10.1007/bf03210847] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
8
|
Patel MM, Goyal BR, Bhadada SV, Bhatt JS, Amin AF. Getting into the brain: approaches to enhance brain drug delivery. CNS Drugs 2009; 23:35-58. [PMID: 19062774 DOI: 10.2165/0023210-200923010-00003] [Citation(s) in RCA: 278] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Being the most delicate organ of the body, the brain is protected against potentially toxic substances by the blood-brain barrier (BBB), which restricts the entry of most pharmaceuticals into the brain. The developmental process for new drugs for the treatment of CNS disorders has not kept pace with progress in molecular neurosciences because most of the new drugs discovered are unable to cross the BBB. The clinical failure of CNS drug delivery may be attributed largely to a lack of appropriate drug delivery systems. Localized and controlled delivery of drugs at their desired site of action is preferred because it reduces toxicity and increases treatment efficiency. The present review provides an insight into some of the recent advances made in the field of brain drug delivery.The various strategies that have been explored to increase drug delivery into the brain include (i) chemical delivery systems, such as lipid-mediated transport, the prodrug approach and the lock-in system; (ii) biological delivery systems, in which pharmaceuticals are re-engineered to cross the BBB via specific endogenous transporters localized within the brain capillary endothelium; (iii) disruption of the BBB, for example by modification of tight junctions, which causes a controlled and transient increase in the permeability of brain capillaries; (iv) the use of molecular Trojan horses, such as peptidomimetic monoclonal antibodies to transport large molecules (e.g. antibodies, recombinant proteins, nonviral gene medicines or RNA interference drugs) across the BBB; and (v) particulate drug carrier systems. Receptor-mediated transport systems exist for certain endogenous peptides, such as insulin and transferrin, enabling these molecules to cross the BBB in vivo.The use of polymers for local drug delivery has greatly expanded the spectrum of drugs available for the treatment of brain diseases, such as malignant tumours and Alzheimer's disease. In addition, various drug delivery systems (e.g. liposomes, microspheres, nanoparticles, nanogels and bionanocapsules) have been used to enhance drug delivery to the brain. Recently, microchips and biodegradable polymers have become important in brain tumour therapy.The intense search for alternative routes of drug delivery (e.g. intranasal drug delivery, convection-enhanced diffusion and intrathecal/intraventricular drug delivery systems) has been driven by the need to overcome the physiological barriers of the brain and to achieve high drug concentrations within the brain. For more than 30 years, considerable efforts have been made to enhance the delivery of therapeutic molecules across the vascular barriers of the CNS. The current challenge is to develop drug delivery strategies that will allow the passage of drug molecules through the BBB in a safe and effective manner.
Collapse
Affiliation(s)
- Mayur M Patel
- Institute of Pharmacy, Nirma University of Science and Technology, Ahmedabad, India
| | | | | | | | | |
Collapse
|
9
|
Dwibhashyam VSNM, Nagappa AN. Strategies for enhanced drug delivery to the central nervous system. Indian J Pharm Sci 2008; 70:145-53. [PMID: 20046703 PMCID: PMC2792490 DOI: 10.4103/0250-474x.41446] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 02/17/2008] [Accepted: 02/23/2008] [Indexed: 11/29/2022] Open
Abstract
Treating central nervous system diseases is very challenging because of the presence of a variety of formidable obstacles that impede drug delivery. Physiological barriers like the blood-brain barrier and blood-cerebrospinal fluid barrier as well as various efflux transporter proteins make the entry of drugs into the central nervous system very difficult. The present review provides a brief account of the blood brain barrier, the P-glycoprotein efflux and various strategies for enhancing drug delivery to the central nervous system.
Collapse
Affiliation(s)
- V. S. N. M. Dwibhashyam
- Pharmacy Practice Department, 4th Floor, Shirdi Sai Baba Cancer Hospital, Manipal-576 104, India
| | - A. N. Nagappa
- TherDose Pharma (P) Ltd., Plot No: 30-32, Survey No. 400, Prasanth Nagar, IE, Hyderabad-500 072, India
| |
Collapse
|
10
|
Lai Y, Drobinskaya I, Kolossov E, Chen C, Linn T. Genetic modification of cells for transplantation. Adv Drug Deliv Rev 2008; 60:146-59. [PMID: 18037530 DOI: 10.1016/j.addr.2007.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 08/02/2007] [Indexed: 01/16/2023]
Abstract
Progress in gene therapy has produced promising results that translate experimental research into clinical treatment. Gene modification has been extensively employed in cell transplantation. The main barrier is an effective gene delivery system. Several viral vectors were utilized in end-stage differentiated cells. Recently, successful applications were described with adenovirus-associated vectors. As an alternative, embryonic stem cell- and stem cell-like systems were established for generation of tissue-specified gene-modified cells. Owing to the feasibility for genetic manipulations and the self-renewing potency of these cells they can be used in a way enabling large-scale in vitro production. This approach offers the establishment of in vitro cell culture systems that will deliver sufficient amounts of highly purified, immunoautologous cells suitable for application in regenerative medicine. In this review, the current technology of gene delivery systems to cells is recapitulated and the latest developments for cell transplantation are discussed.
Collapse
|
11
|
Liu W, Liu Z, Cao X, Cao Z, Xue L, Zhu F, He X, Li W. Recombinant human foamy virus, a novel vector for neurological disorders gene therapy, drives production of GAD in cultured astrocytes. Mol Ther 2007; 15:1834-41. [PMID: 17579580 DOI: 10.1038/sj.mt.6300224] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Human foamy virus (HFV), with its nonpathogenic nature and several unique features for gene transfer, is a promising vector system for neurological disorders gene therapy. The question of whether HFV vectors can be developed for the expression of therapeutic genes in primary astrocytes of the brain may be of interest. First, efficient expression for foreign genes, which is critical for the potentials of HFV-derived vector in gene therapy, was successfully demonstrated in rat-cultured astrocytes by the enhanced green fluorescent protein (EGFP) transduction through an HFV vector bearing an EGFP expression cassette. Second, HFV vectors containing human glutamic acid decarboxylase (GAD) complementary DNA, which encodes an inhibitory neurotransmitter gamma-aminobutyric acid (GABA)-producing enzyme, were used to examine the function of GAD on GABA synthesis in cultured astrocytes. We found that the transduction of GAD vector resulted in isoform-specific expression of GAD, synthesis of a significant amount of GABA and tonical GABA release, and behavioral recovery in rat Parkinson's disease (PD) models. These results suggested that HFV vector had the ability to transduce astrocytes and HFV vector-derived GAD expression in astrocytes provided a potential strategy for the treatment of neurological disorders associated with hyperexcitable or diminished inhibitory activity.
Collapse
Affiliation(s)
- Wanhong Liu
- School of Medicine, Wuhan University, Wuhan, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Pencalet P, Serguera C, Corti O, Privat A, Mallet J, Giménez y Ribotta M. Integration of genetically modified adult astrocytes into the lesioned rat spinal cord. J Neurosci Res 2006; 83:61-7. [PMID: 16294335 DOI: 10.1002/jnr.20697] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Combination of ex vivo gene transfer and cell transplantation is now considered as a potentially useful strategy for the treatment of spinal cord injury. In a perspective of clinical application, autologous transplantation could be an option of choice. We analyzed the fate of adult rat cortical astrocytes genetically engineered with a lentiviral vector transplanted into a lesioned rat spinal cord. Cultures of adult rat cortical astrocytes were infected with an HIV-1-derived vector (TRIP-CMV-GFP) and labeled with the fluorescent dye Hoechst. Transfected and labeled astrocyte suspension was injected at T11 in rats in which spinal cord transection at T7-T8 levels had been carried out 1 week earlier. Six weeks after grafting, the animals were sacrificed and transplants were retrieved either by Hoechst fluorescence or by immunohistochemistry for detection of glial fibrillary acidic protein (GFAP) and vimentin. Grafted astrocytes expressing green fluorescent protein (GFP) were found both at the injection and transection sites. Genetically modified astrocytes thus survived, integrated, and migrated within the host parenchyma when grafted into the completely transected rat spinal cord. In addition, they retained some ability to express the GFP transgene for at least 6 weeks after transplantation. Adult astrocytes infected with lentiviral vectors can therefore be a valuable tool for the delivery of therapeutic factors into the lesioned spinal cord.
Collapse
|
13
|
Hendriks WT, Ruitenberg MJ, Blits B, Boer GJ, Verhaagen J. Viral vector-mediated gene transfer of neurotrophins to promote regeneration of the injured spinal cord. PROGRESS IN BRAIN RESEARCH 2004; 146:451-76. [PMID: 14699980 DOI: 10.1016/s0079-6123(03)46029-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Injuries to the adult mammalian spinal cord often lead to severe damage to both ascending (sensory) pathways and descending (motor) nerve pathways without the perspective of complete functional recovery. Future spinal cord repair strategies should comprise a multi-factorial approach addressing several issues, including optimalization of survival and function of spared central nervous system neurons in partial lesions and the modulation of trophic and inhibitory influences to promote and guide axonal regrowth. Neurotrophins have emerged as promising molecules to augment neuroprotection and neuronal regeneration. Although intracerebroventricular, intrathecal and local protein delivery of neurotrophins to the injured spinal cord has resulted in enhanced survival and regeneration of injured neurons, there are a number of drawbacks to these methods. Viral vector-mediated transfer of neurotrophin genes to the injured spinal cord is emerging as a novel and effective strategy to express neurotrophins in the injured nervous system. Ex vivo transfer of neurotrophic factor genes is explored as a way to bridge lesions cavities for axonal regeneration. Several viral vector systems, based on herpes simplex virus, adenovirus, adeno-associated virus, lentivirus, and moloney leukaemia virus, have been employed. The genetic modification of fibroblasts, Schwann cells, olfactory ensheathing glia cells, and stem cells, prior to implantation to the injured spinal cord has resulted in improved cellular nerve guides. So far, neurotrophic factor gene transfer to the injured spinal cord has led to results comparable to those obtained with direct protein delivery, but has a number of advantages. The steady advances that have been made in combining new viral vector systems with a range of promising cellular platforms for ex vivo gene transfer (e.g., primary embryonic neurons, Schwann cells, olfactory ensheating glia cells and neural stem cells) holds promising perspectives for the development of new neurotrophic factor-based therapies to repair the injured nervous system.
Collapse
Affiliation(s)
- William T Hendriks
- Graduate School for Neurosciences Amsterdam, Department of Neuroregeneration, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
14
|
Kakishita K, Nakao N, Sakuragawa N, Itakura T. Implantation of human amniotic epithelial cells prevents the degeneration of nigral dopamine neurons in rats with 6-hydroxydopamine lesions. Brain Res 2003; 980:48-56. [PMID: 12865158 DOI: 10.1016/s0006-8993(03)02875-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We recently found that human amniotic epithelial (HAE) cells secrete biologically active neurotrophins such as brain-derived neurotrophic factor and neurotrophin-3, both of which exhibit trophic activities on dopamine (DA) neurons. The present study explored whether implantation of HAE cells can be a possible means to deliver trophic factors into the brain to prevent the death of DA neurons in a rat model of Parkinson's disease. We first investigated the ability of HAE cells to produce factors capable of promoting DA cell survival in vitro, and then tested whether HAE cell grafts survive and prevent the death of nigral DA neurons in rats with 6-hydroxydopamine lesions. A treatment with conditioned medium derived from HAE cell cultures enhanced the survival of tyrosine hydroxylase (TH)-immunopositive DA cells in serum-free cultures. The conditioned medium also protected the morphological integrity of TH-positive neurons against toxic insult with 6-hydroxydopamine. HAE cells were grafted into the midbrain of immunosuppressed rats. The rats were then subjected to a unilateral nigrostriatal lesion induced by intrastriatal infusions of 6-hydroxydopamine. HAE cell transplants were found to survive without evidence for overgrowth 2 weeks postgrafting. The number of nigral DA cells, detected with either TH-immunohistochemistry or retrograde labelling with fluorogold, was significantly increased in rats given the grafts as compared to that in control animals without the grafts. The results indicate that HAE cells produce diffusible molecules that can enhance the survival of DA neurons. Although the factors that contribute to the currently observed effects remain to be fully determined, implantation of HAE cells could be a viable strategy to counteract the loss of DA neurons in Parkinson's disease.
Collapse
Affiliation(s)
- Koji Kakishita
- Department of Neurological Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-0012, Japan
| | | | | | | |
Collapse
|
15
|
Lu D, Mahmood A, Chopp M. Biologic Transplantation and Neurotrophin-Induced Neuroplasticity After Traumatic Brain Injury. J Head Trauma Rehabil 2003; 18:357-76. [PMID: 16222130 DOI: 10.1097/00001199-200307000-00006] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE In this review, we analyze progress in the treatment of traumatic brain injury with neurotrophins, growth factors and cell and tissue neurotransplantation. The primary objective of these therapies is to reduce neurologic deficits associated with the trauma by inducing neuroplasticity. These therapies are restorative and not necessarily neuroprotective. MAIN OUTCOME MEASURES An extensive literature on administration of neurotrophics factors and cell and tissue cerebral transplantation is reviewed. The effects of these therapeutic approaches on brain biochemical, molecular, cellular, and tissue responses are summarized. CONCLUSION The cumulative data indicate that cell therapy shows substantial promise in the treatment of neural injury.
Collapse
Affiliation(s)
- Dunyue Lu
- Department of Neurosurgery, Henry Ford Health System, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | | | | |
Collapse
|
16
|
Wang ZH, Ji Y, Shan W, Zeng B, Raksadawan N, Pastores GM, Wisniewski T, Kolodny EH. Therapeutic effects of astrocytes expressing both tyrosine hydroxylase and brain-derived neurotrophic factor on a rat model of Parkinson's disease. Neuroscience 2002; 113:629-40. [PMID: 12150782 DOI: 10.1016/s0306-4522(02)00204-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Tyrosine hydroxylase (TH) and brain-derived neurotrophic factor (BDNF), expressed in normal astrocytes, were used in combination for the treatment of Parkinson's disease (PD) symptoms in a rat model. Normal neonatal rat astrocytes were co-transfected with a vector expressing BDNF (AAVBDNF) and a retroviral vector expressing TH (termed TH-BDNF-DA(+) cells), and then implanted into the striatum of PD rats induced by 6-hydroxydopamine. TH-BDNF-DA(+) cells compensated for a severe insufficiency of endogenous dopaminergic neurons in the PD rats, resulting in a significant improvement of PD symptoms. The decrease in the rotational rate of PD rats implanted with TH-BDNF-DA(+) cells was more marked than that in PD rats implanted with normal astrocytes expressing either TH or BDNF alone (termed TH(+) and BDNF(+) cells, P<0.01 and 0.001, respectively), and suggested a synergistic effect between TH and BDNF. In contrast, the rotational rate was not altered from the baseline in PD rats without treatment or implanted with parental rat astrocytes alone (P>0.05). BDNF protected the dopaminergic neurons from apoptosis induced by 6-hydroxydopamine, and significantly increased the long-term survival of TH-positive cells in the striatum. Our data indicate that the combined use of TH and BDNF has a synergistic therapeutic effect, and is more efficient for the treatment of PD than a single gene therapy using either TH or BDNF alone.
Collapse
Affiliation(s)
- Z H Wang
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The concept of replacing lost dopamine neurons in Parkinson's disease using mesencephalic brain cells from fetal cadavers has been supported by over 20 years of research in animals and over a decade of clinical studies. The ambitious goal of these studies was no less than a molecular and cellular "cure" for Parkinson's disease, other neurodegenerative diseases, and spinal cord injury. Much research has been done in rodents, and a few studies have been done in nonhuman primate models. Early uncontrolled clinical reports were enthusiastic, but the outcome of the first randomized, double blind, controlled study challenged the idea that dopamine replacement cells can cure Parkinson's disease, although there were some significant positive findings. Were the earlier animal studies and clinical reports wrong? Should we give up on the goal? Some aspects of the trial design and implantation methods may have led to lack of effects and to some side effects such as dyskinesias. But a detailed review of clinical neural transplants published to date still suggests that neural transplantation variably reverses some aspects of Parkinson's disease, although differing methods make exact comparisons difficult. While the randomized clinical studies have been in progress, new methods have shown promise for increasing transplant survival and distribution, reconstructing the circuits to provide dopamine to the appropriate targets and with normal regulation. Selected promising new strategies are reviewed that block apoptosis induced by tissue dissection, promote vascularization of grafts, reduce oxidant stress, provide key growth factors, and counteract adverse effects of increased age. New sources of replacement cells and stem cells may provide additional advantages for the future. Full recovery from parkinsonism appears not only to be possible, but a reliable cell replacement treatment may finally be near.
Collapse
Affiliation(s)
- D Eugene Redmond
- Department of Psychiatry, Yale University School of Medicine, USA.
| |
Collapse
|
18
|
Barondeau DP, Kassmann CJ, Tainer JA, Getzoff ED. Structural chemistry of a green fluorescent protein Zn biosensor. J Am Chem Soc 2002; 124:3522-4. [PMID: 11929238 DOI: 10.1021/ja0176954] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We designed a green fluorescent protein mutant (BFPms1) that preferentially binds Zn(II) (enhancing fluorescence intensity) and Cu(II) (quenching fluorescence) directly to a chromophore ligand that resembles a dipyrrole unit of a porphyrin. Crystallographic structure determination of apo, Zn(II)-bound, and Cu(II)-bound BFPms1 to better than 1.5 A resolution allowed us to refine metal centers without geometric restraints, to calculate experimental standard uncertainty errors for bond lengths and angles, and to model thermal displacement parameters anisotropically. The BFPms1 Zn(II) site (KD = 50 muM) displays distorted trigonal bipyrimidal geometry, with Zn(II) binding to Glu222, to a water molecule, and tridentate to the chromophore ligand. In contrast, the BFPms1 Cu(II) site (KD = 24 muM) exhibits square planar geometry similar to metalated porphyrins, with Cu(II) binding to the chromophore chelate and Glu222. The apo structure reveals a large electropositive region near the designed metal insertion channel, suggesting a basis for the measured metal cation binding kinetics. The preorganized tridentate ligand is accommodated in both coordination geometries by a 0.4 A difference between the Zn and Cu positions and by distinct rearrangements of Glu222. The highly accurate metal ligand bond lengths reveal different protonation states for the same oxygen bound to Zn vs Cu, with implications for the observed metal ion specificity. Crystallographic anisotropic thermal factor analysis validates metal ion rigidification of the chromophore in enhancement of fluorescence intensity upon Zn(II) binding. Thus, our high-resolution structures reveal how structure-based design has effectively linked selective metal binding to changes in fluorescent properties. Furthermore, this protein Zn(II) biosensor provides a prototype suitable for further optimization by directed evolution to generate metalloprotein variants with desirable physical or biochemical properties.
Collapse
Affiliation(s)
- David P Barondeau
- Department of Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
19
|
Ericson C, Wictorin K, Lundberg C. Ex vivo and in vitro studies of transgene expression in rat astrocytes transduced with lentiviral vectors. Exp Neurol 2002; 173:22-30. [PMID: 11771936 DOI: 10.1006/exnr.2001.7829] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Implantation of cells genetically modified to express therapeutic genes into the brain has been proposed as a potential treatment for neurodegenerative diseases. In the current study embryonic rat-derived astrocytes were cultured and transduced with a lentiviral vector expressing the reporter gene green fluorescent protein (GFP) and subsequently grafted into the adult rat brain. The proportion of GFP expressing cells was stable, albeit small (1%), at all survival times, up to 6 weeks, the longest time point studied. In parallel in vitro studies, the astrocytes were lentivirally transduced to express either one of the two isoforms of glutamate decarboxylase (GAD(65) or GAD(67)) or glial cell line-derived neurotrophic factor (GDNF). When transducing 293T cells with the two GAD vectors, released GABA could be measured using high-performance liquid chromatography. Further studies of rat astrocytes transduced with the same vectors resulted in a level of GAD activity about 10 times higher than the activity of an intact rat striatum. One hundred thousand astrocytes transduced with LV-GDNF released approximately 27 ng of GDNF per hour. Thus, taken together, our observations provide support for the use of rat astrocytes in ex vivo gene transfer of these proteins in animal models of CNS disorders, e.g., Parkinson's disease or epilepsy.
Collapse
Affiliation(s)
- Cecilia Ericson
- Wallenberg Neuroscience Center, Department of Physiology Sciences, Lund University, S-221 84 Lund, Sweden.
| | | | | |
Collapse
|
20
|
Englund U, Fricker-Gates RA, Lundberg C, Björklund A, Wictorin K. Transplantation of human neural progenitor cells into the neonatal rat brain: extensive migration and differentiation with long-distance axonal projections. Exp Neurol 2002; 173:1-21. [PMID: 11771935 DOI: 10.1006/exnr.2001.7750] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Here we examined the ability of human neural progenitors from the embryonic forebrain, expanded for up to a year in culture in the presence of growth factors, to respond to environmental signals provided by the developing rat brain. After survival times of up to more than a year after transplantation into the striatum, the hippocampus, and the subventricular zone, the cells were analyzed using human-specific antisera and the reporter gene green fluorescent protein (GFP). From grafts implanted in the striatum, the cells migrated extensively, especially within white matter structures. Neuronal differentiation was most pronounced at the striatal graft core, with axonal projections extending caudally along the internal capsule into mesencephalon. In the hippocampus, cells migrated throughout the entire hippocampal formation and into adjacent white matter tracts, with differentiation into neurons both in the dentate gyrus and in the CA1-3 regions. Directed migration along the rostral migratory stream to the olfactory bulb and differentiation into granule cells were observed after implantation into the subventricular zone. Glial differentiation occurred at all three graft sites, predominantly at the injection sites, but also among the migrating cells. A lentiviral vector was used to transduce the cells with the GFP gene prior to grafting. The reporter gene was expressed for at least 15 weeks and the distribution of the gene product throughout the entire cytoplasmic compartment of the expressing cells allowed for a detailed morphological analysis of a portion of the grafted cells. The extensive integration and differentiation of in vitro-expanded human neural progenitor cells indicate that multipotent progenitors are capable of responding in a regionally specific manner to cues present in the developing rat brain.
Collapse
Affiliation(s)
- Ulrica Englund
- Wallenberg Neuroscience Center, Division of Neurobiology, Lund University, S-221 84 Lund, Sweden
| | | | | | | | | |
Collapse
|
21
|
Abstract
The use of fetal astrocytes for gene delivery into brains with neurodegenerative diseases has been suggested. Therefore, the effects of neurotransmitters in the brain on such cells are of interest. The presence of D1(D1A) receptors and the effect of dopamine on a fetal human astrocyte cell line (SVG cells) in vitro were examined. SVG cells expressed D1(D(1A)), but not D5(D1B) receptors, as shown by RT-PCR. Exposure to dopamine, apomorphine, and the specific D1 agonist, SKF-38393, increased glial-derived neurotrophic factor production of SVG cells, as well as intracellular free calcium. Exposure to the specific D1 antagonist, SCH 23390, blocked these effects. Thus, if implanted into a brain region rich in dopamine, or if transfected with the tyrosine hydroxylase gene, fetal astrocytes may serve as paracrine/autocrine cells capable of supplying critical growth factors to diseased brain tissue.
Collapse
Affiliation(s)
- N Kinor
- Department of Life Science, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | | | |
Collapse
|
22
|
Englund U, Ericson C, Rosenblad C, Mandel RJ, Trono D, Wictorin K, Lundberg C. The use of a recombinant lentiviral vector for ex vivo gene transfer into the rat CNS. Neuroreport 2000; 11:3973-7. [PMID: 11192612 DOI: 10.1097/00001756-200012180-00014] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A major obstacle in ex vivo gene transfer has been the loss of transgene expression soon after implantation of the grafted transduced cells. Recently, a lentiviral vector system has been developed which has proven to express high levels of transgenes in vivo after direct injection into the tissue. In this study, we have investigated the use of such a vector for ex vivo gene transfer to the brain. A number of neural cell types were found to be permissive to transduction by the lentiviral vector in vitro and a majority of them expressed the transgene after transplantation to the rat brain. Transgene expression was detected up to 8 weeks post-grafting. These findings suggest that recombinant lentiviral vectors may be used for further development of ex vivo gene therapy protocols to the CNS.
Collapse
Affiliation(s)
- U Englund
- Wallenberg Neuroscience Center, Division of Neurobiology, Lund University, Sweden
| | | | | | | | | | | | | |
Collapse
|
23
|
Pan D, Aronovich E, McIvor RS, Whitley CB. Retroviral vector design studies toward hematopoietic stem cell gene therapy for mucopolysaccharidosis type I. Gene Ther 2000; 7:1875-83. [PMID: 11110421 DOI: 10.1038/sj.gt.3301298] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To optimize a gene transfer system for hematopoietic stem cell gene therapy of patients with mucopolysaccharidosis (MPS) type I, 10 retroviral vectors were constructed to express the human alpha-L-iduronidase (IDUA) cDNA. These vectors were designed to evaluate the potential effects of specific promoters, the addition of selectable markers, and the use of multiple promoters versus an internal ribosome entry site for expression of IDUA and selectable maker genes. The effect of vector design was investigated in primary patient fibroblasts (F(MPS)) or murine fibroblast cell lines; while overall comparison of transgene expression was determined in patients' peripheral blood lymphocytes (PBL(MPS)) and CD34+ progenitors (PBPC(MPS)). We observed that the human PGK promoter introduced the highest IDUA activity per 1% relative transgene frequency in F(MPS). Use of the same promoter to separately regulate both the therapeutic gene and a drug-resistance gene resulted in decreased expression of the unselected gene. Co-selection using bicistronic vectors not only increased the number of transductants, but also elevated transgene expression under selective pressure in transgene-positive progenitors. Bicistronic vector LP1CD overcame down-regulation and practically introduced the highest IDUA level in unselected PBL(MPS) and an intermediate level in PBPC(MPS). These studies provide a better understanding of factors contributing to efficient gene expression in hematopoietic cells.
Collapse
Affiliation(s)
- D Pan
- Department of Pediatrics, and Institute of Human Genetics, University of Minnesota, Minneapolis 55455, USA
| | | | | | | |
Collapse
|
24
|
King LA, Mitrophanous KA, Clark LA, Kim VN, Rohll JB, Kingsman AJ, Colello RJ. Growth factor enhanced retroviral gene transfer to the adult central nervous system. Gene Ther 2000; 7:1103-11. [PMID: 10918476 DOI: 10.1038/sj.gt.3301198] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The use of viral vectors for gene delivery into mammalian cells provides a new approach in the treatment of many human diseases. The first viral vector approved for human clinical trials was murine leukemia virus (MLV), which remains the most commonly used vector in clinical trials to date. However, the application of MLV vectors is limited since MLV requires cells to be actively dividing in order for transduction and therefore gene delivery to occur. This limitation precludes the use of MLV for delivering genes to the adult CNS, where very little cell division is occurring. However, we speculated that this inherent limitation of ML V may be overcome by utilizing the known mitogenic effect of growth factors on cells of the CNS. Specifically, an in vivo application of growth factor to the adult brain, if able to induce cell division, could enhance MLV-based gene transfer to the adult brain. We now show that an exogenous application of basic fibroblast growth factor induces cell division in vivo. Under these conditions, where cells of the adult brain are stimulated to divide, MLV-based gene transfer is significantly enhanced. This novel approach precludes any vector modifications and provides a simple and effective way of delivering genes to cells of the adult brain utilizing MLV-based retroviral vectors.
Collapse
Affiliation(s)
- L A King
- Department of Anatomy, Medical College of Virginia, Virginia Commonwealth University, Richmond 23298-0709, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Eriksson C, Ericson C, Gates MA, Wictorin K. Long-term, EGF-stimulated cultures of attached GFAP-positive cells derived from the embryonic mouse lateral ganglionic eminence: in vitro and transplantation studies. Exp Neurol 2000; 164:184-99. [PMID: 10877929 DOI: 10.1006/exnr.2000.7424] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Long-term attached cultures, prepared from mouse embryonic days 15-17 lateral ganglionic eminence, were grown in a medium including epidermal growth factor and serum, and the survival, differentiation, and migration of cells from either early or late passages were analyzed following transplantation. The cultured cells had the morphology of type I astroglial cells, with the vast majority of the cells immunoreactive for glial fibrillary acidic protein (around 90%), the intermediate filament marker nestin, and also the mouse-specific neural markers M2 and M6. The cultures were kept over 25 passages (7 months). During the first 8 passages, the growth rate gradually declined, but it increased again after passage 9 and thereafter stabilized at values similar to those observed during the initial culture period. After passages 4-6 and 18, cell suspensions were implanted cross-species into the intact or lesioned striatum of adult (passages 4-5 only) or intact striatum of neonatal rats (passages 4-6 or 18). Both early and late passage cells formed M2 (and M6)-positive transplants. In the neonatal recipients, widespread migration was seen from the needle tract throughout most of the striatum, along the internal capsule, and into the globus pallidus. In the adult striatum, the cells remained mostly around the injection tract, or within 0.4-0.6 mm from the graft core. These long-term attached cultures are interesting to compare to nonattached neurosphere cultures, and might also offer a means of propagating relatively pure populations of astroglia-like cells for basic transplantation studies or for use in experimental trials with ex vivo gene transfer.
Collapse
Affiliation(s)
- C Eriksson
- Wallenberg Neuroscience Center, Lund University, Sweden
| | | | | | | |
Collapse
|
26
|
Abstract
The discovery within the past decade that neural stem cells (NSCs) from the developing and adult mammalian brain can be propagated, cloned, and genetically manipulated ex vivo for ultimate transfer back into the CNS has opened the door to a novel means for modifying the CNS environment for experimental and therapeutic purposes. While a great deal of interest has been focused on the properties and promise of this new technology, especially in regard to cellular replacement and gene therapy, this minireview will focus on the recent use of NSCs to study the neuropathogenesis of the murine oncornaviruses. In brief, the use of this NSC-based approach has provided a means for selective reconstitution within the brain, of specific retroviral life cycle events, in order to consider their contribution to the induction of neurodegeneration. Furthermore, by virtue of their ability to disseminate virus within the brain, NSCs have provided a reliable means for assessing the true neurovirulence potential of murine oncornaviruses by directly circumventing a restriction to virus entry into the CNS. Importantly, these experiments have demonstrated that the neurovirulence of oncornaviruses requires late virus life cycle events occurring specifically within microglia, the resident macrophages of the brain. This initial application of NSC biology to the analysis of oncornavirus-CNS interactions may serve as an example for how other questions in viral neuropathogenesis might be addressed in the future.
Collapse
Affiliation(s)
- W P Lynch
- Department of Microbiology/Immunology, Northeastern Ohio Universities College of Medicine, Rootstown 44272-0095, USA.
| | | |
Collapse
|
27
|
Nakao N, Yokote H, Nakai K, Itakura T. Promotion of survival and regeneration of nigral dopamine neurons in a rat model of Parkinson's disease after implantation of embryonal carcinoma-derived neurons genetically engineered to produce glial cell line-derived neurotrophic factor. J Neurosurg 2000; 92:659-70. [PMID: 10761657 DOI: 10.3171/jns.2000.92.4.0659] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The P19 embryonal carcinoma-derived cell line consists of undifferentiated multipotential cells, which irreversibly differentiate into mature neurons after exposure to retinoic acid (RA). In the present study, the authors genetically engineered P19 cells to produce glial cell line-derived neurotrophic factor (GDNF), and grafted the cells in a rat model that had been rendered parkinsonian. METHODS Undifferentiated P19 cells were grown in vitro and transduced with GDNF complementary DNA. The level of GDNF released from the transduced cells was measured using an enzyme-linked immunosorbent assay, and its neurotrophic activities were assessed by testing the effects on rat embryonic dopamine (DA) neurons in culture. After having been exposed to RA for 48 hours and allowed to differentiate into postmitotic neurons, the GDNF gene-transduced cells were implanted into the midbrain of immunosuppressed rats. A unilateral nigrostriatal lesion was then induced by intrastriatal infusions of 6-hydroxydopamine. Immunohistochemical analyses performed 4 weeks postgrafting revealed that the GDNF-producing cells expressed several neuronal markers without evidence of overgrowth. The grafts expressed GDNF protein and prevented the death of nigral DA neurons. Furthermore, the GDNF-producing cells implanted 4 weeks after nigrostriatal lesions restored the expression of tyrosine hydroxylase in injured DA neurons and induced their dendritic sprouting. CONCLUSIONS The results indicate that the P19 cell line transduced with the GDNF gene can stably secrete functional levels of GDNF, even after being converted to postmitotic neurons. Because it is has been established that GDNF exerts trophic effects on DA neurons, the means currently used to deliver GDNF into the brain could be a viable strategy to prevent the death of nigral DA neurons in cases of Parkinson's disease.
Collapse
Affiliation(s)
- N Nakao
- Department of Neurological Surgery, Wakayama Medical College, Japan.
| | | | | | | |
Collapse
|
28
|
|
29
|
Cortez N, Trejo F, Vergara P, Segovia J. Primary astrocytes retrovirally transduced with a tyrosine hydroxylase transgene driven by a glial-specific promoter elicit behavioral recovery in experimental Parkinsonism. J Neurosci Res 2000. [DOI: 10.1002/(sici)1097-4547(20000101)59:1<39::aid-jnr6>3.0.co;2-n] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
30
|
Yadid G, Fitoussi N, Kinor N, Geffen R, Gispan I. Astrocyte line SVG-TH grafted in a rat model of Parkinson's disease. Prog Neurobiol 1999; 59:635-61. [PMID: 10845756 DOI: 10.1016/s0301-0082(99)00013-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The present review describes gene transfer into the brain using extraneuronal cells with an ex vivo approach. The mild immunological reactions in the central nervous system to grafts provided the rationale and empirical basis for brain-transplantation, to replace dying cells, of potential clinical relevance. Fetal human astrocytes were genetically engineered to express tyrosine hydroxylase, the rate-limiting enzyme for the synthesis of catecholamines. These cells were also found to produce constitutively and secrete GDNF and interleukins. Therefore, these cells may prove as a drug-delivery system for the treatment of neurological degenerative conditions such as Parkinson's disease (PD). The field of neuronal reconstruction has reached a critical threshold and there is a need to evaluate the variables that will become critical as the field matures. One of the needs is to characterize the neurochemical alterations in the microenvironment in the context of grafted-host connectivity. This review discusses the functional effects of the pharmacologically-active construct, which consists of astrocytes producing L-DOPA and GDNF. The striatum in PD that lacks the dopaminergic projection from the substantia nigra metabolizes and releases dopamine differently from normal tissue and may react to different factors released by the grafted cells. Moreover, neurochemicals of the host tissue may effect grafted cells as well. An understanding of the way in which these neurochemicals are abnormal in PD and their role in the grafted brain is critical to the improvement of reconstructive strategies using cellular therapeutic strategies.
Collapse
Affiliation(s)
- G Yadid
- Faculty of Life Sciences, Neuropharmacology Section, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | | | |
Collapse
|
31
|
Ourednik V, Ourednik J, Park KI, Snyder EY. Neural stem cells -- a versatile tool for cell replacement and gene therapy in the central nervous system. Clin Genet 1999; 56:267-78. [PMID: 10636444 DOI: 10.1034/j.1399-0004.1999.560403.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In recent years, it has become evident that the developing and even the adult mammalian central nervous system contains a population of undifferentiated, multipotent cell precursors, neural stem cells, the plastic properties of which might be of advantage for the design of more effective therapies for many neurological diseases. This article reviews the recent progress in establishing rodent and human clonal neural stem cell lines, their biological properties, and how these cells can be utilized to a correct variety of defects, with prospects for the near future to harness their behaviour for neural stem cell-based treatment of diseases in humans.
Collapse
Affiliation(s)
- V Ourednik
- Department of Neurology, Harvard Medical School, Children's Hospital, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
32
|
Lynch WP, Sharpe AH, Snyder EY. Neural stem cells as engraftable packaging lines can mediate gene delivery to microglia: evidence from studying retroviral env-related neurodegeneration. J Virol 1999; 73:6841-51. [PMID: 10400782 PMCID: PMC112769 DOI: 10.1128/jvi.73.8.6841-6851.1999] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The induction of spongiform myeloencephalopathy by murine leukemia viruses is mediated primarily by infection of central nervous system (CNS) microglia. In this regard, we have previously shown that CasBrE-induced disease requires late, rather than early, virus replication events in microglial cells (W. P. Lynch et al., J. Virol. 70:8896-8907, 1996). Furthermore, neurodegeneration requires the presence of unique sequences within the viral env gene. Thus, the neurodegeneration-inducing events could result from microglial expression of retroviral envelope protein alone or from the interaction of envelope protein with other viral structural proteins in the virus assembly and maturation process. To distinguish between these possible mechanisms of disease induction, we engineered the engraftable neural stem cell line C17-2 into packaging/producer cells in order to deliver the neurovirulent CasBrE env gene to endogenous CNS cells. This strategy resulted in significant CasBrE env expression within CNS microglia without the appearance of replication competent virus. CasBrE envelope expression within microglia was accompanied by increased expression of activation markers F4/80 and Mac-1 (CD11b) but failed to induce spongiform neurodegenerative changes. These results suggest that envelope expression alone within microglia is not sufficient to induce neurodegeneration. Rather, microglia-mediated disease appears to require neurovirulent Env protein interaction with other viral proteins during assembly or maturation. More broadly, the results presented here prove the efficacy of a novel method by which neural stem cell biology may be harnessed for genetically manipulating the CNS, not only for studying neurodegeneration but also as a paradigm for the disseminated distribution of retroviral vector-transduced genes.
Collapse
Affiliation(s)
- W P Lynch
- Department of Microbiology/Immunology, Northeastern Ohio Universities College of Medicine, Rootstown, Ohio 44272, USA
| | | | | |
Collapse
|
33
|
Park KI, Liu S, Flax JD, Nissim S, Stieg PE, Snyder EY. Transplantation of neural progenitor and stem cells: developmental insights may suggest new therapies for spinal cord and other CNS dysfunction. J Neurotrauma 1999; 16:675-87. [PMID: 10511240 DOI: 10.1089/neu.1999.16.675] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multipotent neural progenitors and stem cells may integrate appropriately into the developing and degenerating central nervous system. They may also be effective in the replacement of genes, cells, and nondiffusible factors in either a widespread or a more circumscribed manner, depending on the therapeutic demands of the clinical situation. In addition, they may be uniquely responsive to some types of neurodegenerative conditions. We believe that these various appealing capabilities are the normal expression of basic biologic properties and attributes of a stem cell. The therapeutic utility of some of those properties is illustrated in this review of ongoing work in our laboratory, particularly with regard to spinal dysfunction. In these examples, we believe we have tapped into a mechanism that underlies a remarkable degree of natural plasticity programmed into the nervous system at the cellular level, and we have now exploited those properties for therapeutic ends.
Collapse
Affiliation(s)
- K I Park
- Department of Neurology, Harvard Medical School, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
The ability to isolate multipotential neuroepithelial precursor cells from the mammalian nervous system provides exciting perspectives for the in vitro analysis of early nervous system development and the generation of donor cells for neural repair. New models are needed to study the properties of these cells in vivo. Neural chimeras have revealed a remarkable degree of plasticity in the developmental potential of neuroepithelial precursor cells. Following transplantation into the cerebral ventricle of embryonic hosts, precursors derived from various brain regions and developmental stages participate in host brain development and undergo region-specific differentiation into neurons and glia. These findings indicate that in the developing nervous system, migration and differentiation of neural precursors cells are regulated to a large extent by extrinsic signals. Neural chimeras composed of genetically modified cells will permit the study of the molecular mechanisms underlying these guidance cues, which may eventually be exploited for cell replacement strategies in the adult brain. A key problem in neural transplantation is the availability of suitable donor tissue. Neural chimeras composed of embryonic stem (ES) cell-derived neurons and glia depict ES cells as a versatile and virtually unlimited donor source for neural repair. Generation of interspecies neural chimeras composed of human and rodent cells facilitates the translation of these advances into clinical strategies for human nervous system repair.
Collapse
Affiliation(s)
- O Brüstle
- Department of Neuropathology, University of Bonn Medical Center, Germany.
| |
Collapse
|
35
|
Vescovi AL, Snyder EY. Establishment and properties of neural stem cell clones: plasticity in vitro and in vivo. Brain Pathol 1999; 9:569-98. [PMID: 10416994 PMCID: PMC8098170 DOI: 10.1111/j.1750-3639.1999.tb00542.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The study of the basic physiology of the neural precursors generated during brain development is driven by two inextricably linked goals. First, such knowledge is instrumental to our understanding of how the high degree of cellular complexity of the mature central nervous system (CNS) is generated, and how to dissect the steps of proliferation, fate commitment, and differentiation that lead early pluripotent neural progenitors to give rise to mature CNS cells. Second, it is hoped that the isolation, propagation, and manipulation of brain precursors and, particularly, of multipotent neural stem cells (NSCs), will lead to therapeutic applications in neurological disorders. The debate is still open concerning the most appropriate definition of a stem cell and on how it is best identified, characterized, and manipulated. By adopting an operational definition of NSCs, we review some of the basic findings in this area and elaborate on their potential therapeutic applications. Further, we discuss recent evidence from our two groups that describe, based on that rigorous definition, the isolation and propagation of clones of NSCs from the human fetal brain and illustrate how they have begun to show promise for neural cell replacement and molecular support therapy in models of degenerative CNS diseases. The extensive propagation and engraftment potential of human CNS stem cells may, in the not-too-distant-future, be directed towards genuine clinical therapeutic ends, and may open novel and multifaceted strategies for redressing a variety of heretofore untreatable CNS dysfunctions.
Collapse
Affiliation(s)
- Angelo L. Vescovi
- Laboratory of Neuropharmacology, National Neurological Institute C. Besta, Via Celoria 11, 20133 Milan, Italy
| | - Evan Y. Snyder
- Departments of Neurology (Division of Neuroscience), Pediatrics (Division of Newborn Medicine), Neurosurgery (Division of Neuroscience Research), Harvard Medical School, Children's Hospital, Boston, MA USA
| |
Collapse
|
36
|
Transplants of fibroblasts genetically modified to express BDNF promote regeneration of adult rat rubrospinal axons and recovery of forelimb function. J Neurosci 1999. [PMID: 10341240 DOI: 10.1523/jneurosci.19-11-04370.1999] [Citation(s) in RCA: 344] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adult mammalian CNS neurons do not normally regenerate their severed axons. This failure has been attributed to scar tissue and inhibitory molecules at the injury site that block the regenerating axons, a lack of trophic support for the axotomized neurons, and intrinsic neuronal changes that follow axotomy, including cell atrophy and death. We studied whether transplants of fibroblasts genetically engineered to produce brain-derived neurotrophic factor (BDNF) would promote rubrospinal tract (RST) regeneration in adult rats. Primary fibroblasts were modified by retroviral-mediated transfer of a DNA construct encoding the human BDNF gene, an internal ribosomal entry site, and a fusion gene of lacZ and neomycin resistance genes. The modified fibroblasts produce biologically active BDNF in vitro. These cells were grafted into a partial cervical hemisection cavity that completely interrupted one RST. One and two months after lesion and transplantation, RST regeneration was demonstrated with retrograde and anterograde tracing techniques. Retrograde tracing with fluorogold showed that approximately 7% of RST neurons regenerated axons at least three to four segments caudal to the transplants. Anterograde tracing with biotinylated dextran amine revealed that the RST axons regenerated through and around the transplants, grew for long distances within white matter caudal to the transplant, and terminated in spinal cord gray matter regions that are the normal targets of RST axons. Transplants of unmodified primary fibroblasts or Gelfoam alone did not elicit regeneration. Behavioral tests demonstrated that recipients of BDNF-producing fibroblasts showed significant recovery of forelimb usage, which was abolished by a second lesion that transected the regenerated axons.
Collapse
|
37
|
Ourednik V, Ourednik J, Park KI, Snyder EY. Neural stem cells - a versatile tool for cell replacement and gene therapy in the central nervous system. Clin Genet 1999. [DOI: 10.1034/j.1399-0004.2000.57si01.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Ljungberg MC, Stern G, Wilkin GP. Survival of genetically engineered, adult-derived rat astrocytes grafted into the 6-hydroxydopamine lesioned adult rat striatum. Brain Res 1999; 816:29-37. [PMID: 9878682 DOI: 10.1016/s0006-8993(98)01061-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Astrocytes are potentially useful as vehicles for gene transfer into the CNS. As endogenous CNS cells, they possess secretory mechanisms and can be grown in vitro. We have developed an animal model of this system using autologous astrocyte grafts in Fischer 344 rats. Cultured cells were infected with an adenoviral vector containing the reporter gene lacZ in vitro and then grafted into the striatum of adult Fischer 344 rats previously lesioned with 6-OHDA. Survival of the cells and activity of the beta-galactosidase protein were followed for up to 21 days after injection. The grafted cells were shown to survive throughout the experimental period although the expression of transgene was reduced with time. If long-term expression of therapeutically active substances can be achieved, grafts of adult-derived astrocytes genetically engineered using recombinant adenoviral vectors could be employed in the treatment of Parkinson's disease and other neurological disorders.
Collapse
Affiliation(s)
- M C Ljungberg
- Biochemistry Department, Imperial College, Exhibition Road, London SW7 2AZ, UK
| | | | | |
Collapse
|
39
|
Abstract
The neuronal ceroid lipofuscinoses (NCLs, also known as Batten disease) are the most common childhood neurodegenerative disease. They are a group of inherited neurodegenerative disorders characterized by the accumulation of autofluorescent storage material in many cell types. Clinical features include seizures, psychomotor deterioration, and blindness, the ages and order of onset of which differ for each NCL type. An increasing number of subtypes caused by mutations in different genes are now recognized. With the advent of molecular genetics the basic genetic defect underlying each NCL phenotype is being determined, thus shedding light on the molecular basis of the NCLs and opening the way for the development of effective treatment. Four genes have been identified to date. The function of two of these is known and suggests that the primary defect in the NCLs lies in lysosomal proteolysis, the first example of this type of disease. However, since the function of the other two genes remains elusive, and at least four more genes remain to be identified, the molecular basis underlying the NCLs may be more complex than originally predicted.
Collapse
Affiliation(s)
- S E Mole
- Department of Paediatrics, University College London Medical School, United Kingdom
| |
Collapse
|
40
|
Goldman SA, Nedergaard M, Crystal RG, Fraser RA, Goodman R, Harrison-Restelli C, Jiang J, Keyoung HM, Leventhal C, Pincus DW, Shahar A, Wang S. Neural precursors and neuronal production in the adult mammalian forebrain. Ann N Y Acad Sci 1997; 835:30-55. [PMID: 9616760 DOI: 10.1111/j.1749-6632.1997.tb48616.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- S A Goldman
- Department of Neurology and Neuroscience, Cornell University Medical College, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|